1
|
Glaschke S, Dobrovolny HM. Spatiotemporal spread of oncolytic virus in a heterogeneous cell population. Comput Biol Med 2024; 183:109235. [PMID: 39369544 DOI: 10.1016/j.compbiomed.2024.109235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Oncolytic (cancer-killing) virus treatment is a promising new therapy for cancer, with many viruses currently being tested for their ability to eradicate tumors. One of the major stumbling blocks to the development of this treatment modality has been preventing spread of the virus to non-cancerous cells. Our recent ability to manipulate RNA and DNA now allows for the possibility of creating designer viruses specifically targeted to cancer cells, thereby significantly reducing unwanted side effects in patients. In this study, we use a partial differential equation model to determine the characteristics of a virus needed to contain spread of an oncolytic virus within a spherical tumor and prevent it from spreading to non-cancerous cells outside the tumor. We find that oncolytic viruses that have different infection rates or different cell death rates in cancer and non-cancerous cells can be made to stay within the tumor. We find that there is a minimum difference in infection rates or cell death rates that will contain the virus and that this threshold value depends on the growth rate of the cancer. Identification of these types of thresholds can help researchers develop safer strains of oncolytic viruses allowing further development of this promising treatment.
Collapse
Affiliation(s)
- Sabrina Glaschke
- Institute of Physics, Universitat Kassel, Kassel, Germany; Department of Physics & Astronomy, Texas Christian University, Fort Worth, TX, USA
| | - Hana M Dobrovolny
- Department of Physics & Astronomy, Texas Christian University, Fort Worth, TX, USA.
| |
Collapse
|
2
|
Patel S, Salaman SD, Kapoor DU, Yadav R, Sharma S. Latest developments in biomaterial interfaces and drug delivery: challenges, innovations, and future outlook. Z NATURFORSCH C 2024:znc-2024-0208. [PMID: 39566511 DOI: 10.1515/znc-2024-0208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/03/2024] [Indexed: 11/22/2024]
Abstract
An ideal drug carrier system should demonstrate optimal payload and release characteristics, thereby ensuring prolonged therapeutic index while minimizing adverse effects. The field of drug delivery has undergone significant advancements, particularly within the last two decades, owing to the revolutionary impact of biomaterials. The use of biomaterials presents significant due to their biocompatibility and biodegradability, which must be addressed in order to achieve effective drug delivery. The properties of the biomaterial and its interface are primarily influenced by their physicochemical attributes, physiological barriers, cellular trafficking, and immunomodulatory effects. By attuning these barriers, regulating the physicochemical properties, and masking the immune system's response, the bio interface can be effectively modulated, leading to the development of innovative supramolecular structures with enhanced effectiveness. With a comprehensive understanding of these technologies, there is a growing demand for repurposing existing drugs for new therapeutic indications within this space. This review aims to provide a substantial body of evidence showcasing the productiveness of biomaterials and their interface in drug delivery, as well as methods for mitigating and modulating barriers and physicochemical properties along with an examination of future prospects in this field.
Collapse
Affiliation(s)
- Saraswati Patel
- Department of Pharmacology, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai, 602105, Tamil Nadu, India
| | - Samsi D Salaman
- Apollo College of Pharmacy, Mevaloorkuppam, Kanchipuram, 602105, Tamil Nadu, India
| | - Devesh U Kapoor
- Dr. Dayaram Patel Pharmacy College, Sardar Baug, Station Road, 394601 Bardoli, Gujarat, India
| | - Richa Yadav
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, P.O., Rajasthan, 304022, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, P.O., Rajasthan, 304022, India
| |
Collapse
|
3
|
Zhang N, Zhou J, Li S, Cai W, Ru B, Hu J, Liu W, Liu X, Tong X, Zheng X. Advances in Nanoplatforms for Immunotherapy Applications Targeting the Tumor Microenvironment. Mol Pharm 2024; 21:410-426. [PMID: 38170627 DOI: 10.1021/acs.molpharmaceut.3c00846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Cancer immunotherapy is a treatment method that activates or enhances the autoimmune response of the body to fight tumor growth and metastasis, has fewer toxic side effects and a longer-lasting efficacy than radiotherapy and chemotherapy, and has become an important means for the clinical treatment of cancer. However, clinical results from immunotherapy have shown that most patients lack responsiveness to immunotherapy and cannot benefit from this treatment strategy. The tumor microenvironment (TME) plays a critical role in the response to immunotherapy. The TME typically prevents effective lymphocyte activation, reducing their infiltration, and inhibiting the infiltration of effector T cells. According to the characteristic differences between the TME and normal tissues, various nanoplatforms with TME targeting and regulation properties have been developed for more precise regulation of the TME and have the ability to codeliver a variety of active pharmaceutical ingredients, thereby reducing systemic toxicity and improving the therapeutic effect of antitumor. In addition, the precise structural design of the nanoplatform can integrate specific functional motifs, such as surface-targeted ligands, degradable backbones, and TME stimulus-responsive components, into nanomedicines, thereby reshaping the tumor microenvironment, improving the body's immunosuppressive state, and enhancing the permeability of drugs in tumor tissues, in order to achieve controlled and stimulus-triggered release of load cargo. In this review, the physiological characteristics of the TME and the latest research regarding the application of TME-regulated nanoplatforms in improving antitumor immunotherapy will be described. Furthermore, the existing problems and further applications perspectives of TME-regulated platforms for cancer immunotherapy will also be discussed.
Collapse
Affiliation(s)
- Nannan Zhang
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Junyu Zhou
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Shun Li
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Wenjun Cai
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Bin Ru
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Jiaqi Hu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Wenlong Liu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xuanxi Liu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiangmin Tong
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xiaoyan Zheng
- Department of Clinical Laboratory, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang 324000, China
| |
Collapse
|
4
|
Meng S, Du H, Li X, Zheng X, Zhao P, Yuan Z, Huang S, Zhao Y, Dai L. An Adjuvant Micelle-Based Multifunctional Nanosystem for Tumor Immunotherapy by Remodeling Three Types of Immunosuppressive Cells. ACS NANO 2024; 18:3134-3150. [PMID: 38236616 DOI: 10.1021/acsnano.3c08792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Immunotherapy is restricted by a complex tumor immunosuppressive microenvironment (TIM) and low drug delivery efficiency. Herein, a multifunctional adjuvant micelle nanosystem (PPD/MPC) integrated with broken barriers and re-education of three classes of immune-tolerant cells is constructed for cancer immunotherapy. The nanosystem significantly conquers the penetration barrier via the weakly acidic tumor microenvironment-responsive size reduction and charge reversal strategy. The detached core micelle MPC could effectively be internalized by tumor-associated macrophages (TAMs), tumor-infiltrating dendritic cells (TIDCs), and myeloid-derived suppressor cells (MDSCs) via mannose-mediated targeting endocytosis and electrostatic adsorption pathways, promoting the re-education of immunosuppressive cells for allowing them to reverse from pro-tumor to antitumor phenotypes by activating TLR4/9 pathways. This process in turn leads to the remodeling of TIM. In vitro and in vivo studies collectively indicate that the adjuvant micelle-based nanosystem not only relieves the intricate immune tolerance and remodels TIM via reprogramming the three types of immunosuppressive cells and regulating the secretion of relevant cytokines/immunity factors but also strengthens immune response and evokes immune memory, consequently suppressing the tumor growth and metastasis.
Collapse
Affiliation(s)
- Siyu Meng
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, People's Republic of China
| | - Huiping Du
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, People's Republic of China
| | - Xiang Li
- School of Life Science, Northwestern Polytechnical University, Xian 710072, China
| | - Xinmin Zheng
- School of Life Science, Northwestern Polytechnical University, Xian 710072, China
| | - Pan Zhao
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, People's Republic of China
| | - Zhang Yuan
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, People's Republic of China
| | - Shaohui Huang
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing 101499, China
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Liangliang Dai
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, People's Republic of China
| |
Collapse
|
5
|
Rana P, Singh C, Kaushik A, Saleem S, Kumar A. Recent advances in stimuli-responsive tailored nanogels for cancer therapy; from bench to personalized treatment. J Mater Chem B 2024; 12:382-412. [PMID: 38095136 DOI: 10.1039/d3tb02650g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
To improve the quality of health in a personalized manner, better control over pharmacologically relevant cargo formulation, organ-specific targeted delivery, and on-demand release of therapeutic agents is crucial. Significant work has been put into designing and developing revolutionary nanotherapeutics approaches for the effective monitoring and personalized treatment of disease. Nanogel (NG) has attracted significant interest because of its tremendous potential in cancer therapy and its environmental stimuli responsiveness. NG is considered a next-generation delivery technology due to its benefits like as size tunability, high loading, stimuli responsiveness, prolonged drug release via in situ gelling mechanisms, stability, and its potential to provide personalized therapy from the investigation of human genes and the genes in various types of cancers and its association with a selective anticancer drug. Stimuli-responsive NGs can be used as smart nanomedicines to detect and treat cancer and can be tuned as personalized medicine as well. This comprehensive review article's major objectives include the challenges of NGs' clinical translation for cancer treatment as well as its early preclinical successes and prospects.
Collapse
Affiliation(s)
- Prinsy Rana
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
- M. M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala-133207, Haryana, India
| | - Charan Singh
- Department of Pharmaceutical Sciences, School of Sciences, Hemvati Nandan Bahuguna Garhwal University (A Central University), Srinagar, Uttarakhand-246174, India
| | - Ajeet Kaushik
- NanoBiotech Lab, Department of Environmental Engineering, Florida Polytechnic University (FPU), Lakeland, FL, 33805-8531, USA
- School of Engineering, University of Petroleum and Energy Studies, Dehradun 248007, India
| | - Shakir Saleem
- Department of Public Health, College of Health Sciences, Saudi Electronic University, P. O. Box 93499, Riyadh 11673, Saudi Arabia
| | - Arun Kumar
- Department of Pharmacy, School of Health Sciences, Central University of South Bihar, Gaya-824209, India.
| |
Collapse
|
6
|
Li R, Hao Y, Pan W, Wang W, Min Y. Monophosphoryl lipid A-assembled nanovaccines enhance tumor immunotherapy. Acta Biomater 2023; 171:482-494. [PMID: 37708924 DOI: 10.1016/j.actbio.2023.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 08/12/2023] [Accepted: 09/10/2023] [Indexed: 09/16/2023]
Abstract
Therapeutic cancer nanovaccines can induce strong antitumor immunity and establish long-term immune memory and have shown potential for curing tumors in some clinical trials. However, weak immunogenicity and safety concerns of nanocarriers limit the clinical translation of some therapeutic nanovaccines. Here, we developed minimal-component cancer nanovaccines, monophosphoryl lipid A (MPLA)-assembled nanovaccines (MANs), that could facilitate the clinical application of nanovaccines. The MANs were formed by protein antigens extracted from chemotherapy-induced tumor cell cultures and the amphiphilic immune adjuvant MPLA. Compared with free chemotherapy-induced antigens, MANs can activate the Toll-like receptor 4 (TLR4)-mediated signalling pathway and promote adaptive immunity against tumor antigens. Mechanistic analysis indicated that MANs induced antigen capture of DCs and promoted the activation of DCs and T cells, thereby optimizing the ratio of CD8+ T/Tregs in tumors and facilitating the transformation of the tumor immune microenvironment (TIME) from "cold" to "hot". In a CT26 colorectal cancer model, MANs+αPD-1 significantly improved the efficacy of αPD-1 treatment. Our work offers a strategy for designing minimal-component cancer nanovaccines with potential clinical benefits. STATEMENT OF SIGNIFICANCE: To address the weak immunogenicity of cancer vaccines and the safety concerns of nanocarriers, we prepared MPLA-assembled nanovaccines (MANs) using chemotherapy induced antigens and the immune adjuvant MPLA to promote cancer vaccines to clinical practice. MANs effectively internalized tumor antigens and induced DC maturation, indicating that the initial anti-tumor response had been activated. MANs+αPD-1 induced APCs, CD8+ T cells and memory T cells with positive anti-tumor effects to migrate to tumor tissue, thus leading to the transformation of the tumor immune microenvironment from "cold" to "hot". At the animal level, the combination of MANs and αPD-1 exerted synergistic effects and significantly enhanced tumor immunotherapy. Therefore, the treatment regimen of MANs+αPD-1 has potential clinical benefits.
Collapse
Affiliation(s)
- Rui Li
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Yuhao Hao
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Wen Pan
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Wei Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Yuanzeng Min
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China; Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Hefei, 230026, China; CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
7
|
Li Y, Luo Y, Hou L, Huang Z, Wang Y, Zhou S. Antigen-Capturing Dendritic-Cell-Targeting Nanoparticles for Enhanced Tumor Immunotherapy Based on Photothermal-Therapy-Induced In Situ Vaccination. Adv Healthc Mater 2023; 12:e2202871. [PMID: 37276021 DOI: 10.1002/adhm.202202871] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 05/30/2023] [Indexed: 06/07/2023]
Abstract
In situ vaccines have revolutionized immunotherapy as they can stimulate tumor-specific immune responses, with the cancer being the antigen source. However, the heterogeneity of tumor antigens and insufficient dendritic cells (DCs) activation result in low cancer immunogenicity and hence poor vaccine response. Herein, a new in situ vaccine composed of acid-responsive liposome-coated polydopamine (PDA) nanoparticles modified with mannose and loaded with resiquimod (R848) is designed to promote the efficacy of immunotherapy. The in situ vaccine can actively target the tumor site based on the decomposition of the liposome, while the PDA nanoparticles promote photothermal therapy and capture the immunogenic cell-death-induced tumor-associated antigens based on the adsorption effect of dopamine-mimetic mussels. The PDA nanoparticles, which are modified with a mannose ligand, target the DCs and release R848 for activated antigen presentation. As a result, the in situ vaccine not only effectively activates the maturation of the DCs but also significantly enhances their effect on cytotoxic T lymphocyte cells. Furthermore, the vaccine effectively inhibits the distant recurrence and metastasis of tumors via long-term immune memory effects. Therefore, the in situ vaccine provides a potential strategy for improving the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Yingmin Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Yang Luo
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Lamei Hou
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Zhengjie Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Yi Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| |
Collapse
|
8
|
Wei X, Yu CY, Wei H. Application of Cyclodextrin for Cancer Immunotherapy. Molecules 2023; 28:5610. [PMID: 37513483 PMCID: PMC10384645 DOI: 10.3390/molecules28145610] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Tumor immunotherapy, compared with other treatment strategies, has the notable advantage of a long-term therapeutic effect for preventing metastasis and the recurrence of tumors, thus holding great potential for the future of advanced tumor therapy. However, due to the poor water solubility of immune modulators and immune escape properties of tumor cells, the treatment efficiency of immunotherapy is usually significantly reduced. Cyclodextrin (CD) has been repeatedly highlighted to be probably one of the most investigated building units for cancer therapy due to its elegant integration of an internal hydrophobic hollow cavity and an external hydrophilic outer surface. The application of CD for immunotherapy provides new opportunities for overcoming the aforementioned obstacles. However, there are few published reviews, to our knowledge, summarizing the use of CD for cancer immunotherapy. For this purpose, this paper provides a comprehensive summary on the application of CD for immunotherapy with an emphasis on the role, function, and reported strategies of CD in mediating immunotherapy. This review summarizes the research progress made in using CD for tumor immunotherapy, which will facilitate the generation of various CD-based immunotherapeutic delivery systems with superior anticancer efficacy.
Collapse
Affiliation(s)
- Xiaojie Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| |
Collapse
|
9
|
Shen K, Ke S, Chen B, Zhang T, Wang H, Lv J, Gao W. Identification and validation of biomarkers for epithelial-mesenchymal transition-related cells to estimate the prognosis and immune microenvironment in primary gastric cancer by the integrated analysis of single-cell and bulk RNA sequencing data. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:13798-13823. [PMID: 37679111 DOI: 10.3934/mbe.2023614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
BACKGROUND The epithelial-mesenchymal transition (EMT) is associated with gastric cancer (GC) progression and immune microenvironment. To better understand the heterogeneity underlying EMT, we integrated single-cell RNA-sequencing (scRNA-seq) data and bulk sequencing data from GC patients to evaluate the prognostic utility of biomarkers for EMT-related cells (ERCs), namely, cancer-associated fibroblasts (CAFs) and epithelial cells (ECs). METHODS scRNA-seq data from primary GC tumor samples were obtained from the Gene Expression Omnibus (GEO) database to identify ERC marker genes. Bulk GC datasets from the Cancer Genome Atlas (TCGA) and GEO were used as training and validation sets, respectively. Differentially expressed markers were identified from the TCGA database. Univariate Cox, least-absolute shrinkage, and selection operator regression analyses were performed to identify EMT-related cell-prognostic genes (ERCPGs). Kaplan-Meier, Cox regression, and receiver-operating characteristic (ROC) curve analyses were adopted to evaluate the prognostic utility of the ERCPG signature. An ERCPG-based nomogram was constructed by integrating independent prognostic factors. Finally, we evaluated the correlations between the ERCPG signature and immune-cell infiltration and verified the expression of ERCPG prognostic signature genes by in vitro cellular assays. RESULTS The ERCPG signature was comprised of seven genes (COL4A1, F2R, MMP11, CAV1, VCAN, FKBP10, and APOD). Patients were divided into high- and low-risk groups based on the ERCPG risk scores. Patients in the high-risk group showed a poor prognosis. ROC and calibration curves suggested that the ERCPG signature and nomogram had a good prognostic utility. An immune cell-infiltration analysis suggested that the abnormal expression of ERCPGs induced the formation of an unfavorable tumor immune microenvironment. In vitro cellular assays showed that ERCPGs were more abundantly expressed in GC cell lines compared to normal gastric tissue cell lines. CONCLUSIONS We constructed and validated an ERCPG signature using scRNA-seq and bulk sequencing data from ERCs of GC patients. Our findings support the estimation of patient prognosis and tumor treatment in future clinical practice.
Collapse
Affiliation(s)
- Kaiyu Shen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shuaiyi Ke
- Department of Internal Medicine, XianJu People's Hospital, XianJu 317399, China
| | - Binyu Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Tiantian Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Hongtai Wang
- Department of General Surgery, XianJu People' Hospital, XianJu 317399, China
| | - Jianhui Lv
- Department of General Surgery, XianJu People' Hospital, XianJu 317399, China
| | - Wencang Gao
- Department of Oncology, Zhejiang Chinese Medical University, Hangzhou 310005, China
| |
Collapse
|
10
|
Li Q, Liu X, Yan C, Zhao B, Zhao Y, Yang L, Shi M, Yu H, Li X, Luo K. Polysaccharide-Based Stimulus-Responsive Nanomedicines for Combination Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206211. [PMID: 36890780 DOI: 10.1002/smll.202206211] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 02/09/2023] [Indexed: 06/08/2023]
Abstract
Cancer immunotherapy is a promising antitumor approach, whereas nontherapeutic side effects, tumor microenvironment (TME) intricacy, and low tumor immunogenicity limit its therapeutic efficacy. In recent years, combination immunotherapy with other therapies has been proven to considerably increase antitumor efficacy. However, achieving codelivery of the drugs to the tumor site remains a major challenge. Stimulus-responsive nanodelivery systems show controlled drug delivery and precise drug release. Polysaccharides, a family of potential biomaterials, are widely used in the development of stimulus-responsive nanomedicines due to their unique physicochemical properties, biocompatibility, and modifiability. Here, the antitumor activity of polysaccharides and several combined immunotherapy strategies (e.g., immunotherapy combined with chemotherapy, photodynamic therapy, or photothermal therapy) are summarized. More importantly, the recent progress of polysaccharide-based stimulus-responsive nanomedicines for combination cancer immunotherapy is discussed, with the focus on construction of nanomedicine, targeted delivery, drug release, and enhanced antitumor effects. Finally, the limitations and application prospects of this new field are discussed.
Collapse
Affiliation(s)
- Qiuxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Xing Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Chunmei Yan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Bolin Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Yuxin Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Lu Yang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Mingyi Shi
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hua Yu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao SAR, 999078, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Kaipei Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| |
Collapse
|
11
|
Kalami A, Shahgolzari M, Khosroushahi AY, Fiering S. Combining in situ vaccination and immunogenic apoptosis to treat cancer. Immunotherapy 2023; 15:367-381. [PMID: 36852419 DOI: 10.2217/imt-2022-0137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
Immunization approaches are designed to stimulate the immune system and eliminate the tumor. Studies indicate that cancer immunization combined with certain chemotherapeutics and immunostimulatory agents can improve outcomes. Chemotherapeutics-based immunogenic cell death makes the tumor more recognizable by the immune system. In situ vaccination (ISV) utilizes established tumors as antigen sources and directly applies an immune adjuvant to the tumor to reverse a cold tumor microenvironment to a hot one. Immunogenic cell death and ISV highlight for the immune system the tumor antigens that are recognizable by immune cells and support a T-cell attack of the tumor cells. This review presents the concept of immunogenic apoptosis and ISV as a powerful platform for cancer immunization.
Collapse
Affiliation(s)
- Arman Kalami
- Biotechnology Research Center, Student Research Committee, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Shahgolzari
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Yari Khosroushahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Steven Fiering
- Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth & Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| |
Collapse
|
12
|
CAR-T cells for cancer immunotherapy. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
|
13
|
Liu XZ, Wen ZJ, Li YM, Sun WR, Hu XQ, Zhu JZ, Li XY, Wang PY, Pedraz JL, Lee JH, Kim HW, Ramalingam M, Xie S, Wang R. Bioengineered Bacterial Membrane Vesicles with Multifunctional Nanoparticles as a Versatile Platform for Cancer Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:3744-3759. [PMID: 36630299 DOI: 10.1021/acsami.2c18244] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Inducing immunogenic cell death (ICD) is a critical strategy for enhancing cancer immunotherapy. However, inefficient and risky ICD inducers along with a tumor hypoxia microenvironment seriously limit the immunotherapy efficacy. Non-specific delivery is also responsible for this inefficiency. In this work, we report a drug-free bacteria-derived outer membrane vesicle (OMV)-functionalized Fe3O4-MnO2 (FMO) nanoplatform that realized neutrophil-mediated targeted delivery and photothermally enhanced cancer immunotherapy. In this system, modification of OMVs derived from Escherichia coli enhanced the accumulation of FMO NPs at the tumor tissue through neutrophil-mediated targeted delivery. The FMO NPs underwent reactive decomposition in the tumor site, generating manganese and iron ions that induced ICD and O2 that regulated the tumor hypoxia environment. Moreover, OMVs are rich in pathogen-associated pattern molecules that can overcome the tumor immunosuppressive microenvironment and effectively activate immune cells, thereby enhancing specific immune responses. Photothermal therapy (PTT) caused by MnO2 and Fe3O4 can not only indirectly stimulate systemic immunity by directly destroying tumor cells but also promote the enrichment of neutrophil-equipped nanoparticles by enhancing the inflammatory response at the tumor site. Finally, the proposed multi-modal treatment system with targeted delivery capability realized effective tumor immunotherapy to prevent tumor growth and recurrence.
Collapse
Affiliation(s)
- Xin Zheng Liu
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai264003, PR China
- Shandong Laboratory of Advanced Materials and Green Manufacturing, Yantai264000, People's Republic of China
| | - Zhi Juan Wen
- Binzhou Medical University Hospital, Binzhou256603PR China
| | - Yun Meng Li
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai264003, PR China
| | - Wan Ru Sun
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai264003, PR China
| | - Xiao Qian Hu
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai264003, PR China
| | - Jia Zhi Zhu
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai264003, PR China
| | - Xin Yu Li
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai264003, PR China
| | - Ping Yu Wang
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai264003, People's Republic of China
| | - José Luis Pedraz
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006Vitoria-Gasteiz, Spain
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, 28029Madrid, Spain
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan31116, Republic of Korea
- Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan31116, Republic of Korea
- Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan31116, Republic of Korea
| | - Murugan Ramalingam
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan31116, Republic of Korea
- Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan31116, Republic of Korea
- School of Basic Medical Sciences, Chengdu University, Chengdu610106, People's Republic of China
- Department of Metallurgical and Materials Engineering, Atilim University, Ankara06830, Turkey
| | - Shuyang Xie
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai264003, People's Republic of China
| | - Ranran Wang
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai264003, PR China
- Shandong Laboratory of Advanced Materials and Green Manufacturing, Yantai264000, People's Republic of China
| |
Collapse
|
14
|
Zhang X, Khan S, Wei R, Zhang Y, Liu Y, Wee Yong V, Xue M. Application of nanomaterials in the treatment of intracerebral hemorrhage. J Tissue Eng 2023; 14:20417314231157004. [PMID: 37032735 PMCID: PMC10074624 DOI: 10.1177/20417314231157004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/28/2023] [Indexed: 04/05/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is a non-traumatic hemorrhage caused by the rupture of blood vessels in the brain parenchyma, with an acute mortality rate of 30%‒40%. Currently, available treatment options that include surgery are not promising, and new approaches are urgently needed. Nanotechnology offers new prospects in ICH because of its unique benefits. In this review, we summarize the applications of various nanomaterials in ICH. Nanomaterials not only enhance the therapeutic effects of drugs as delivery carriers but also contribute to several facets after ICH such as repressing detrimental neuroinflammation, resisting oxidative stress, reducing cell death, and improving functional deficits.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Suliman Khan
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Ruixue Wei
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Voon Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
15
|
Han S, Wu J. Development of a Lysine-Based Poly(ester amide) Library with High Biosafety and a Finely Tunable Structure for Spatiotemporal-Controlled Protein Delivery. ACS APPLIED MATERIALS & INTERFACES 2022; 14:55944-55956. [PMID: 36503257 DOI: 10.1021/acsami.2c16492] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
With the fast growth of protein therapeutics, efficient, precise, and universal delivery platforms are highly required. However, very few reports have discussed the progress of precisely spatiotemporal-controlled protein delivery. Therefore, a mini library of well-designed amino acid-based poly(ester amide)s derived from lysine (Lys-aaPEAs) has been developed. Lys-aaPEAs can interact with and encapsulate proteins into nanocomplexes via electrostatic interactions. The chemical structure of Lys-aaPEAs can be finely tuned by changing the type and molar ratio of the monomers. Studies of structure-function relationships reveal that the carbon chain length of diacid/diol segments, hydrophilicity, and electrical properties affect the polymer-protein interaction, cell-material interaction, and, therefore, the outcome of protein delivery. By modulating the structures of Lys-aaPEAs, the delivery systems could present customized physiochemical and biological properties and perform time- and space-specific protein release and delivery without causing any systematic toxicity. The screened systems exhibited prolonged hypoglycemic activity and superior biosafety in vivo, using insulin as a model protein and a mouse model bearing type 1 diabetes mellitus (T1DM). This work establishes a novel lysine-based polymer platform for spatiotemporal-controlled protein delivery and offers a paradigm of precise structure-function controllability for designing the next generation of polymers.
Collapse
Affiliation(s)
- Shuyan Han
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, P. R. China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Jun Wu
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, P. R. China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510006, P. R. China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, P. R. China
| |
Collapse
|
16
|
Su L, Hao Y, Li R, Pan W, Ma X, Weng J, Min Y. Red blood cell-based vaccines for ameliorating cancer chemoimmunotherapy. Acta Biomater 2022; 154:401-411. [PMID: 36241013 DOI: 10.1016/j.actbio.2022.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/23/2022] [Accepted: 10/03/2022] [Indexed: 12/14/2022]
Abstract
Immune checkpoint blockade (ICB) therapy has shown promising antitumor effects, but its immune response rate remains unsatisfactory. In recent years, chemotherapy has been proven to have synergistic effects with ICB therapy because some chemotherapeutic agents can enhance the immunogenicity of tumor cells by inducing immunogenic cell death (ICD). However, it cannot be ignored that chemotherapy often shows limited therapeutic efficacy due to high cytotoxicity, drug resistance, and some other side effects. Herein, we report a strategy to improve cancer immunotherapy by utilizing red blood cell-based vaccines (RBC-vaccines) where chemotherapy-induced tumor antigens (cAgs) are anchored onto red blood cells (RBCs) via the EDC/NHS-mediated amine coupling reaction. In this work, RBC-vaccines administered subcutaneously are primarily devoured by dendritic cells (DCs) and significantly improve the efficacy of αPD-1 (anti-programmed cell death 1) treatment by increasing the infiltration of intratumoral CD8+ and CD4+ T cells and elevating the intratumoral ratio of CD8+ T cells to regulatory T cells in the CT-26 colon cancer model. Finally, based on the rejection of tumor rechallenge in cured mice, the combination therapy of RBC-vaccines and αPD-1 can induce the expansion of memory T cells and thereby establish a long-term antitumor immune response. Taken together, the proposed RBC-vaccines have great potential to improve chemoimmunotherapy. STATEMENT OF SIGNIFICANCE: Immunotherapy, especially immune checkpoint blockade therapy, has made great contributions to the treatment of some advanced cancers. Unfortunately, the great majority of patients with cancer do not benefit from immunotherapy. To enhance the response rate of immunotherapy, we developed red blood cell-based vaccines (RBC-vaccines) against cancers where antigens were harvested from chemotherapy-treated cancer cells and then attached to erythrocytes via covalent surface modification. Such RBC-vaccines could provide a wide variety of tumor antigens and damage-associated molecular patterns without the use of any extra ingredients to trigger a stronger antitumor immune response. More importantly, the combination of RBC-vaccines with PD-1 blockade could significantly improve the efficacy of cancer immunotherapy and induce durable antitumor immunity.
Collapse
Affiliation(s)
- Lanhong Su
- Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Yuhao Hao
- Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Rui Li
- Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Wen Pan
- Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Xiaopeng Ma
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Jianping Weng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yuanzeng Min
- Department of Chemistry, University of Science and Technology of China, Hefei, China; The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China; CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
17
|
Han S, Wu J. Three-dimensional (3D) scaffolds as powerful weapons for tumor immunotherapy. Bioact Mater 2022; 17:300-319. [PMID: 35386452 PMCID: PMC8965033 DOI: 10.1016/j.bioactmat.2022.01.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 02/07/2023] Open
Abstract
Though increasing understanding and remarkable clinical successes have been made, enormous challenges remain to be solved in the field of cancer immunotherapy. In this context, biomaterial-based immunomodulatory strategies are being developed to boost antitumor immunity. For the local immunotherapy, macroscale biomaterial scaffolds with 3D network structures show great superiority in the following aspects: facilitating the encapsulation, localized delivery, and controlled release of immunotherapeutic agents and even immunocytes for more efficient immunomodulation. The concentrating immunomodulation in situ could minimize systemic toxicities, but still exert abscopal effects to harness the power of overall anticancer immune response for eradicating malignancy. To promote such promising immunotherapies, the design requirements of macroscale 3D scaffolds should comprehensively consider their physicochemical and biological properties, such as porosity, stiffness, surface modification, cargo release kinetics, biocompatibility, biodegradability, and delivery modes. To date, increasing studies have focused on the relationships between these parameters and the biosystems which will guide/assist the 3D biomaterial scaffolds to achieve the desired immunotherapeutic outcomes. In this review, by highlighting some recent achievements, we summarized the latest advances in the development of various 3D scaffolds as niches for cancer immunotherapy. We also discussed opportunities, challenges, current trends, and future perspectives in 3D macroscale biomaterial scaffold-assisted local treatment strategies. More importantly, this review put more efforts to illustrate how the 3D biomaterial systems affect to modulate antitumor immune activities, where we discussed how significant the roles and behaviours of 3D macroscale scaffolds towards in situ cancer immunotherapy in order to direct the design of 3D immunotherapeutic.
Collapse
Affiliation(s)
- Shuyan Han
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen, 518057, China
| | - Jun Wu
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen, 518057, China
| |
Collapse
|
18
|
Xu J, Cao W, Wang P, Liu H. Tumor-Derived Membrane Vesicles: A Promising Tool for Personalized Immunotherapy. Pharmaceuticals (Basel) 2022; 15:ph15070876. [PMID: 35890175 PMCID: PMC9318328 DOI: 10.3390/ph15070876] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor-derived membrane vesicles (TDMVs) are non-invasive, chemotactic, easily obtained characteristics and contain various tumor-borne substances, such as nucleic acid and proteins. The unique properties of tumor cells and membranes make them widely used in drug loading, membrane fusion and vaccines. In particular, personalized vectors prepared using the editable properties of cells can help in the design of personalized vaccines. This review focuses on recent research on TDMV technology and its application in personalized immunotherapy. We elucidate the strengths and challenges of TDMVs to promote their application from theory to clinical practice.
Collapse
Affiliation(s)
- Jiabin Xu
- School of Stomatology, Xuzhou Medical University, Xuzhou 221004, China; (J.X.); (P.W.)
- Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Wenqiang Cao
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd., Jinan University, Zhuhai 519000, China;
| | - Penglai Wang
- School of Stomatology, Xuzhou Medical University, Xuzhou 221004, China; (J.X.); (P.W.)
- Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Hong Liu
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd., Jinan University, Zhuhai 519000, China;
- Correspondence:
| |
Collapse
|
19
|
Gupta M, Chandan K, Sarwat M. Natural Products and their Derivatives as Immune Check Point Inhibitors: Targeting Cytokine/Chemokine Signalling in Cancer. Semin Cancer Biol 2022; 86:214-232. [PMID: 35772610 DOI: 10.1016/j.semcancer.2022.06.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/15/2022] [Accepted: 06/24/2022] [Indexed: 11/29/2022]
Abstract
Cancer immunotherapy is the new generation and widely accepted form of tumour treatment. It is, however, associated with exclusive challenges which include organ-specific inflammation, and single-target strategies. Therefore, approaches that can enhance the efficiency of existing immunotherapies and expand their indications are required for the further development of immunotherapy. Natural products and medicines are stated to have this desired effect on cancer immunotherapy (adoptive immune-cells therapy, cancer vaccines, and immune-check point inhibitors). They refurbish the immunosuppressed tumour microenvironment, which is the primary location of interaction of tumour cells with the host immune system. Various immune cell subsets, via interaction with cytokine/chemokine receptors, are recruited into this microenvironment, and these subsets have roles in tumour progression and treatment responsiveness. This review summarises cytokine/chemokine signalling, types of cancer immunotherapy and the herbal medicine-derived natural products targeting cytokine/chemokines and immune checkpoints. These natural compounds possess immunomodulatory activities and exert their anti-tumour effect by either blocking the interaction or modulating the expression of the proteins linked with immune checkpoint signaling pathways. Some compounds also show a synergistic effect in combination with existing monoclonal antibody drugs to reverse the tumour microenvironment. Additionally, we have also reported some studies about the derivatives and formulations used to overcome the limitations of natural forms. This review can provide important insights for directing future research.
Collapse
Affiliation(s)
- Meenakshi Gupta
- Amity Institute of Pharmacy, Amity University, Noida-201313, Uttar Pradesh, India
| | - Kumari Chandan
- Amity Institute of Pharmacy, Amity University, Noida-201313, Uttar Pradesh, India
| | - Maryam Sarwat
- Amity Institute of Pharmacy, Amity University, Noida-201313, Uttar Pradesh, India.
| |
Collapse
|
20
|
Tumor-Associated Inflammation: The Tumor-Promoting Immunity in the Early Stages of Tumorigenesis. J Immunol Res 2022; 2022:3128933. [PMID: 35733919 PMCID: PMC9208911 DOI: 10.1155/2022/3128933] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/12/2022] [Indexed: 12/12/2022] Open
Abstract
Tumorigenesis is a multistage progressive oncogenic process caused by alterations in the structure and expression level of multiple genes. Normal cells are continuously endowed with new capabilities in this evolution, leading to subsequent tumor formation. Immune cells are the most important components of inflammation, which is closely associated with tumorigenesis. There is a broad consensus in cancer research that inflammation and immune response facilitate tumor progression, infiltration, and metastasis via different mechanisms; however, their protumor effects are equally important in tumorigenesis at earlier stages. Previous studies have demonstrated that during the early stages of tumorigenesis, certain immune cells can promote the formation and proliferation of premalignant cells by inducing DNA damage and repair inhibition, releasing trophic/supporting signals, promoting immune escape, and activating inflammasomes, as well as enhance the characteristics of cancer stem cells. In this review, we focus on the potential mechanisms by which immune cells can promote tumor initiation and promotion in the early stages of tumorigenesis; furthermore, we discuss the interaction of the inflammatory environment and protumor immune cells with premalignant cells and cancer stem cells, as well as the possibility of early intervention in tumor formation by targeting these cellular mechanisms.
Collapse
|
21
|
Two-dimensional nanomaterials for tumor microenvironment modulation and anticancer therapy. Adv Drug Deliv Rev 2022; 187:114360. [PMID: 35636568 DOI: 10.1016/j.addr.2022.114360] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/02/2022] [Accepted: 05/23/2022] [Indexed: 12/28/2022]
Abstract
The development of two-dimensional (2D) nanomaterials for cancer therapy has attracted increasing attention due to their high specific surface area, unique ultrathin structure, electronic and photonic properties. For biomedical applications, investigations into the family of 2D materials have been sparked by graphene and its derivatives. Many 2D nanomaterials, including layered double hydroxides, transition metal dichalcogenides, nitrides and carbonitrides, black phosphorus nanosheets, and metal-organic framework nanosheets, are extensively explored as cancer theranostic platforms. In addition to the high drug loading, 2D nanomaterials are featured with improved physiological properties of drugs, prolonged blood circulation, and increased tumor accumulation and bioavailability. As a consequence, 2D nanomaterials have been widely examined in pre-clinical tumor therapy, particularly through the tumor microenvironment (TME) modulation. This review summarizes recent progresses in developing 2D nanomaterials for TME modulating-based cancer diagnosis and therapy. It is anticipated that this review will benefit researchers to obtain a deeper understanding of interactions between 2D nanomaterials and TME components and develop rational and reliable 2D nanomedicines for pre/clinical cancer theranostics.
Collapse
|
22
|
Le TMD, Yoon AR, Thambi T, Yun CO. Polymeric Systems for Cancer Immunotherapy: A Review. Front Immunol 2022; 13:826876. [PMID: 35273607 PMCID: PMC8902250 DOI: 10.3389/fimmu.2022.826876] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/24/2022] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy holds enormous promise to create a new outlook of cancer therapy by eliminating tumors via activation of the immune system. In immunotherapy, polymeric systems play a significant role in improving antitumor efficacy and safety profile. Polymeric systems possess many favorable properties, including magnificent biocompatibility and biodegradability, structural and component diversity, easy and controllable fabrication, and high loading capacity for immune-related substances. These properties allow polymeric systems to perform multiple functions in immunotherapy, such as immune stimulants, modifying and activating T cells, delivery system for immune cargos, or as an artificial antigen-presenting cell. Among diverse immunotherapies, immune checkpoint inhibitors, chimeric antigen receptor (CAR) T cell, and oncolytic virus recently have been dramatically investigated for their remarkable success in clinical trials. In this report, we review the monotherapy status of immune checkpoint inhibitors, CAR-T cell, and oncolytic virus, and their current combination strategies with diverse polymeric systems.
Collapse
Affiliation(s)
- Thai Minh Duy Le
- Department of Bioengineering, College of Engineering, Hanayang University, Seoul, South Korea
| | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanayang University, Seoul, South Korea.,Institute of Nano Science and Technology (INST), Hanayang University, Seoul, South Korea.,Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, South Korea
| | - Thavasyappan Thambi
- Department of Bioengineering, College of Engineering, Hanayang University, Seoul, South Korea
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanayang University, Seoul, South Korea.,Institute of Nano Science and Technology (INST), Hanayang University, Seoul, South Korea.,Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, South Korea.,GeneMedicine CO., Ltd., Seoul, South Korea
| |
Collapse
|
23
|
Xie R, Yuan M, Jiang Y. The Pan-Cancer Crosstalk Between the EFNA Family and Tumor Microenvironment for Prognosis and Immunotherapy of Gastric Cancer. Front Cell Dev Biol 2022; 10:790947. [PMID: 35309935 PMCID: PMC8924469 DOI: 10.3389/fcell.2022.790947] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/08/2022] [Indexed: 12/24/2022] Open
Abstract
Background: EFNA1-5 have important physiological functions in regulating tumorigenesis and metastasis. However, correlating EFNA genes in the tumor immune microenvironment (TIME), and the prognosis of patients with gastric cancer remains to be determined. Methods: Using public databases, the expression of EFNA1-5 in pan-cancer and gastric cancer was comprehensively analyzed using UCSC Xena, the Oncomine dataset and UALCAN. We further completed survival analysis by Kaplan-Meier plotter to evaluate the prognosis of the high and low expression groups of the EFNAs gene in patients with gastric cancer. The TIMER tool was used to reveal the correlation between immune cell infiltration and genes of interest. Spearman correlation was used to find an association between the EFNA genes and tumor stem cells, TIME, microsatellite instability (MSI) or tumor mutational burden (TMB). We also used cBioportal, GeneMANIA and STRINGS to explore the types of changes in these genes and the protein interactions. Finally, we described the TIME based on QUANTISEQ algorithm, predicted the relationship between the EFNA genes and half-maximal inhibitory concentration (IC50), and analyzed the relationship between the EFNA family genes and immune checkpoints. Results: The expression of EFNA1, EFNA3, EFNA4, and EFNA5 was elevated in pan-cancer. Compared with normal adjacent tissues, EFNA1, EFNA3, and EFNA4 were up-regulated in gastric cancer. In terms of the influence on the survival of patients, the expression of EFNA3 and EFNA4 were related to overall survival (OS) and disease-free survival (DFS) for patients with gastric cancer. High expression of EFNA5 often predicted poor OS and DFS. In gastric cancer, the expression of EFNA3 and EFNA4 showed a significant negative correlation with B cells. The higher the expression of EFNA5, the higher the abundance of B cells, CD4+T cells and macrophages. CD8+T cells, dendritic cells infiltration and EFNA1-4 expression were negatively correlated. The infiltration of CD4+T cells, macrophages and neutrophils was negatively correlated with the expression of EFNA1, EFNA3, and EFNA4. TMB and MSI were positively correlated with EFNA3/EFNA4 expression. In the tumor microenvironment and drug sensitivity, EFNA3/4/5 also showed a significant correlation. In addition, we explored the relationship between the EFNA family genes and the immune microenvironment (B cells, M2 macrophages, monocytes, CD8+ T cells, regulatory T cells, myeloid dendritic cells, natural killer cells, non-regulatory CD4+ T cells), immune checkpoint (PDCD1, PDCD1LG2, CD274, CTLA4), and IC50 of common chemotherapeutic drugs for gastric cancer (5-fluorouracil, cisplatin, docetaxel and gemcitabine). Conclusions: Our study provides new ideas for tumor treatment and prognosis from the perspective of TIME, and nominates EFNA1-5 to become potential therapeutic targets for gastric cancer.
Collapse
Affiliation(s)
- Rongrong Xie
- Department of Radiotherapy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengping Yuan
- Department of Gastroenterology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiyan Jiang
- Department of Medical Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
24
|
Zhuang Y, Han S, Fang Y, Huang H, Wu J. Multidimensional transitional metal-actuated nanoplatforms for cancer chemodynamic modulation. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214360] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Yang L, Li F, Cao Y, Liu Q, Jing G, Niu J, Sun F, Qian Y, Wang S, Li A. Multifunctional silica nanocomposites prime tumoricidal immunity for efficient cancer immunotherapy. J Nanobiotechnology 2021; 19:328. [PMID: 34663354 PMCID: PMC8524820 DOI: 10.1186/s12951-021-01073-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 10/04/2021] [Indexed: 12/18/2022] Open
Abstract
The tumor immune microenvironment (TIME) has been demonstrated to be the main cause of cancer immunotherapy failure in various malignant tumors, due to poor immunogenicity and existence of immunosuppressive factors. Thus, establishing effective treatments for hostile TIME remodeling has considerable potential to enhance immune response rates for durable tumor growth retardation. This study aims to develop a novel nanocomposite, polyethyleneimine-modified dendritic mesoporous silica nanoparticles loaded with microRNA-125a (DMSN-PEI@125a) to synergistically enhance immune response and immunosuppression reversion, ultimately generating a tumoricidal environment. Our results showed that DMSN-PEI@125a exhibited excellent ability in cellular uptake by murine macrophages and the cervical cancer cell line TC-1, repolarization of tumor associated macrophages (TAMs) to M1 type in a synergistic manner, and promotion of TC-1 immunogenic death. Intratumor injection of DMSN-PEI@125a facilitated the release of more damage-related molecular patterns and enhanced the infiltration of natural killer and CD8+ T cells. Meanwhile, repolarized TAMs could function as a helper to promote antitumor immunity, thus inhibiting tumor growth in TC-1 mouse models in a collaborative manner. Collectively, this work highlights the multifunctional roles of DMSN-PEI@125a in generating an inflammatory TIME and provoking antitumor immunity, which may serve as a potential agent for cancer immunotherapy.
Collapse
Affiliation(s)
- Linnan Yang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China.,Central Laboratory, First Affiliated Hospital, Anhui Medical University, Hefei, People's Republic of China
| | - Feng Li
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| | - Yongsheng Cao
- The Second Department of Urology, Anhui Provincial Children's Hospital, Hefei, People's Republic of China
| | - Qiang Liu
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| | - Guoxin Jing
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| | - Jintong Niu
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| | - Feiyue Sun
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| | - Yechang Qian
- Department of Respiratory Disease, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, People's Republic of China.
| | - Shilong Wang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China.
| | - Ang Li
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China.
| |
Collapse
|
26
|
Wu Z, Guan Q, Han X, Liu X, Li L, Qiu L, Qian Z, Zhou S, Wang X, Zhang H. A novel prognostic signature based on immune-related genes of diffuse large B-cell lymphoma. Aging (Albany NY) 2021; 13:22947-22962. [PMID: 34610582 PMCID: PMC8544299 DOI: 10.18632/aging.203587] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/18/2021] [Indexed: 11/25/2022]
Abstract
Diffuse large B-cell lymphoma (DLBCL) presents a great clinical challenge and has a poor prognosis, with immune-related genes playing a crucial role. We aimed to develop an immune-related prognostic signature for improving prognosis prediction in DLBCL. Samples from the GSE31312 dataset were randomly allocated to discovery and internal validation cohorts. Univariate Cox, random forest, LASSO regression and multivariate Cox analyses were utilized to develop a prognostic signature, which was verified in the internal validation cohort, entire validation cohort and external validation cohort (GSE10846). The tumor microenvironment was investigated using the CIBERSORT and ESTIMATE tools. Gene set enrichment analysis (GSEA) was further applied to analyze the entire GSE31312 cohort. We identified four immune-related genes (CD48, IL1RL, PSDM3, RXFP3) significantly associated with overall survival. Based on discovery and validation cohort analyses, this four-gene signature could classify patients into high- and low-risk groups, with significantly different prognoses. Activated memory CD4 T cells and activated dendritic cells were significantly decreased in the high-risk group, and these patients had lower immune scores. GSEA revealed enrichment of signaling pathways, such as T cell receptor, antigen receptor-mediated, antigen processing and presentation of peptide antigen via MHC class I, in the low-risk group. In conclusion, a robust signature based on four immune-related genes was successfully constructed for predicting prognosis in DLBCL patients.
Collapse
Affiliation(s)
- Zizheng Wu
- Departments of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Qingpei Guan
- Departments of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Xue Han
- Departments of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Xianming Liu
- Departments of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Lanfang Li
- Departments of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Lihua Qiu
- Departments of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Zhengzi Qian
- Departments of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Shiyong Zhou
- Departments of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Xianhuo Wang
- Departments of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| | - Huilai Zhang
- Departments of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Sino-US Center for Lymphoma and Leukemia Research, Tianjin 300060, China
| |
Collapse
|
27
|
Thangam R, Patel KD, Kang H, Paulmurugan R. Advances in Engineered Polymer Nanoparticle Tracking Platforms towards Cancer Immunotherapy-Current Status and Future Perspectives. Vaccines (Basel) 2021; 9:vaccines9080935. [PMID: 34452059 PMCID: PMC8402739 DOI: 10.3390/vaccines9080935] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/03/2021] [Accepted: 08/13/2021] [Indexed: 12/17/2022] Open
Abstract
Engineering polymeric nanoparticles for their shape, size, surface chemistry, and functionalization using various targeting molecules has shown improved biomedical applications for nanoparticles. Polymeric nanoparticles have created tremendous therapeutic platforms, particularly applications related to chemo- and immunotherapies in cancer. Recently advancements in immunotherapies have broadened this field in immunology and biomedical engineering, where "immunoengineering" creates solutions to target translational science. In this regard, the nanoengineering field has offered the various techniques necessary to manufacture and assemble multifunctional polymeric nanomaterial systems. These include nanoparticles functionalized using antibodies, small molecule ligands, targeted peptides, proteins, and other novel agents that trigger and encourage biological systems to accept the engineered materials as immune enhancers or as vaccines to elevate therapeutic functions. Strategies to engineer polymeric nanoparticles with therapeutic and targeting molecules can provide solutions for developing immune vaccines via maintaining the receptor storage in T- and B cells. Furthermore, cancer immunotherapy using polymeric nanomaterials can serve as a gold standard approach for treating primary and metastasized tumors. The current status of the limited availability of immuno-therapeutic drugs highlights the importance of polymeric nanomaterial platforms to improve the outcomes via delivering anticancer agents at localized sites, thereby enhancing the host immune response in cancer therapy. This review mainly focuses on the potential scientific enhancements and recent developments in cancer immunotherapies by explicitly discussing the role of polymeric nanocarriers as nano-vaccines. We also briefly discuss the role of multifunctional nanomaterials for their therapeutic impacts on translational clinical applications.
Collapse
Affiliation(s)
- Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea; (K.D.P.); (H.K.)
- Institute for High Technology Materials and Devices, Korea University, Seoul 02841, Korea
- Correspondence: (R.T.); (R.P.)
| | - Kapil D. Patel
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea; (K.D.P.); (H.K.)
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Korea; (K.D.P.); (H.K.)
- Institute for High Technology Materials and Devices, Korea University, Seoul 02841, Korea
- Department of Biomicrosystem Technology, Korea University, Seoul 02841, Korea
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
- Correspondence: (R.T.); (R.P.)
| |
Collapse
|
28
|
Wang D, Yao Y, Xiao Y, Chen X, Hu J, Yang X. Ultrasound responsive erythrocyte membrane-derived hybrid nanovesicles with controlled drug release for tumor therapy. NANOSCALE 2021; 13:9945-9951. [PMID: 34057169 DOI: 10.1039/d1nr01916c] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
An ultrasound responsive erythrocyte membrane-derived hybrid nanovesicle drug delivery system (DOX/HMME@FA-NL) is constructed by the membrane fusion functionalization strategy for controlled drug release and enhanced tumor therapy. The reliability and effectiveness of the membrane fusion strategy are confirmed through characterization of the particle size and zeta potential, Förster energy resonance transfer and fluorescence co-localization analyses. The DOX/HMME@FA-NL could be triggered for reactive oxygen species (ROS) generation under ultrasound stimulation. And the unsaturated phospholipids in DOX/HMME@FA-NL can be oxidized by ROS, leading to the destruction of the structure of the hybrid membrane to achieve the controlled release of drugs, thereby enhancing their tumor cell killing effect. Besides, the linkage of the folate targeting group also enhances the tumor targeting ability of DOX/HMME@FA-NL. H22 tumor-bearing mice were intravenously injected with DOX/HMME@FA-NL and treated with ultrasound, they achieved better than expected tumor sonodynamic response treatment effects.
Collapse
Affiliation(s)
- Dongdong Wang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| | | | | | | | | | | |
Collapse
|
29
|
4-1BBL as a Mediator of Cross-Talk between Innate, Adaptive, and Regulatory Immunity against Cancer. Int J Mol Sci 2021; 22:ijms22126210. [PMID: 34207500 PMCID: PMC8227424 DOI: 10.3390/ijms22126210] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 01/25/2023] Open
Abstract
The ability of tumor cells to evade the immune system is one of the main challenges we confront in the fight against cancer. Multiple strategies have been developed to counteract this situation, including the use of immunostimulant molecules that play a key role in the anti-tumor immune response. Such a response needs to be tumor-specific to cause as little damage as possible to healthy cells and also to track and eliminate disseminated tumor cells. Therefore, the combination of immunostimulant molecules and tumor-associated antigens has been implemented as an anti-tumor therapy strategy to eliminate the main obstacles confronted in conventional therapies. The immunostimulant 4-1BBL belongs to the tumor necrosis factor (TNF) family and it has been widely reported as the most effective member for activating lymphocytes. Hence, we will review the molecular, pre-clinical, and clinical applications in conjunction with tumor-associated antigens in antitumor immunotherapy, as well as the main molecular pathways involved in this association.
Collapse
|
30
|
Akkın S, Varan G, Bilensoy E. A Review on Cancer Immunotherapy and Applications of Nanotechnology to Chemoimmunotherapy of Different Cancers. Molecules 2021; 26:3382. [PMID: 34205019 PMCID: PMC8199882 DOI: 10.3390/molecules26113382] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 12/19/2022] Open
Abstract
Clinically, different approaches are adopted worldwide for the treatment of cancer, which still ranks second among all causes of death. Immunotherapy for cancer treatment has been the focus of attention in recent years, aiming for an eventual antitumoral effect through the immune system response to cancer cells both prophylactically and therapeutically. The application of nanoparticulate delivery systems for cancer immunotherapy, which is defined as the use of immune system features in cancer treatment, is currently the focus of research. Nanomedicines and nanoparticulate macromolecule delivery for cancer therapy is believed to facilitate selective cytotoxicity based on passive or active targeting to tumors resulting in improved therapeutic efficacy and reduced side effects. Today, with more than 55 different nanomedicines in the market, it is possible to provide more effective cancer diagnosis and treatment by using nanotechnology. Cancer immunotherapy uses the body's immune system to respond to cancer cells; however, this may lead to increased immune response and immunogenicity. Selectivity and targeting to cancer cells and tumors may lead the way to safer immunotherapy and nanotechnology-based delivery approaches that can help achieve the desired success in cancer treatment.
Collapse
Affiliation(s)
- Safiye Akkın
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkey;
| | - Gamze Varan
- Department of Vaccine Technology, Hacettepe University Vaccine Institute, 06100 Ankara, Turkey;
| | - Erem Bilensoy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkey;
| |
Collapse
|
31
|
Giustarini G, Pavesi A, Adriani G. Nanoparticle-Based Therapies for Turning Cold Tumors Hot: How to Treat an Immunosuppressive Tumor Microenvironment. Front Bioeng Biotechnol 2021; 9:689245. [PMID: 34150739 PMCID: PMC8207137 DOI: 10.3389/fbioe.2021.689245] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Nanotechnologies are rapidly increasing their role in immuno-oncology in line with the need for novel therapeutic strategies to treat patients unresponsive to chemotherapies and immunotherapies. The tumor immune microenvironment (TIME) has emerged as critical for tumor classification and patient stratification to design better treatments. Notably, the tumor infiltration of effector T cells plays a crucial role in antitumor responses and has been identified as the primary parameter to define hot, immunosuppressed, excluded, and cold tumors. Organic and inorganic nanoparticles (NPs) have been applied as carriers of new targeted therapies to turn cold or altered (i.e., immunosuppressed or excluded) tumors into more therapeutically responsive hot tumors. This mini-review discusses the significant advances in NP-based approaches to turn immunologically cold tumors into hot ones.
Collapse
Affiliation(s)
- Giulio Giustarini
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Andrea Pavesi
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology, and Research (ASTAR), Singapore, Singapore
| | - Giulia Adriani
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| |
Collapse
|
32
|
Lima BV, Oliveira MJ, Barbosa MA, Gonçalves RM, Castro F. Immunomodulatory potential of chitosan-based materials for cancer therapy: a systematic review of in vitro, in vivo and clinical studies. Biomater Sci 2021; 9:3209-3227. [PMID: 33949372 DOI: 10.1039/d0bm01984d] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Chitosan (Ch) has recently been used in different studies as a vaccine adjuvant with an ability to modulate the tumor microenvironment (TME). This systematic review aims to elucidate the added value of using Ch-based therapies for immunotherapeutic strategies in cancer treatment, through the exploration of different Ch-based formulations, their capacity to modulate immune cells in vitro and in vivo, and their translational potential for clinical settings. A systematic review was conducted on PubMed, following both inclusion and exclusion steps. Original articles which focused on the immunomodulatory role of Ch-based formulations in the TME were included, as well as its usage as a delivery vehicle for other immunomodulatory molecules. This review illustrates the added value of Ch-based systems to reshape the TME, through the modulation of immune cells using different Ch formulations, namely solutions, films, gels, microneedles and nanoparticles. Generally, Ch-based formulations increase the recruitment and proliferation of cells associated with pro-inflammatory abilities and decrease cells which exert anti-inflammatory activities. These effects correlated with a decreased tumor weight, reduced metastases, reversion of the immunosuppressive TME and increased survival in vivo. Overall, Ch-based formulations present the potential for immunotherapy in cancer. Nevertheless, clinical translation remains challenging, since the majority of the studies use Ch in formulations with other components, implicating that some of the observed effects could result from the combination of the individual effects. More studies on the use of different Ch-based formulations, complementary to standardization and disclosure of the Ch properties used are required to improve the immunomodulatory effects of Ch-based formulations in cancer.
Collapse
Affiliation(s)
- Beatriz V Lima
- i3S - Institute of Research and Innovation in Health, University of Porto, Porto, Portugal. and INEB - Institute of Biomedical Engineering, University of Porto, Porto, Portugal and ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Maria J Oliveira
- i3S - Institute of Research and Innovation in Health, University of Porto, Porto, Portugal. and INEB - Institute of Biomedical Engineering, University of Porto, Porto, Portugal and ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Mário A Barbosa
- i3S - Institute of Research and Innovation in Health, University of Porto, Porto, Portugal. and INEB - Institute of Biomedical Engineering, University of Porto, Porto, Portugal and ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Raquel M Gonçalves
- i3S - Institute of Research and Innovation in Health, University of Porto, Porto, Portugal. and INEB - Institute of Biomedical Engineering, University of Porto, Porto, Portugal and ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Flávia Castro
- i3S - Institute of Research and Innovation in Health, University of Porto, Porto, Portugal. and INEB - Institute of Biomedical Engineering, University of Porto, Porto, Portugal
| |
Collapse
|
33
|
Zi L, Zhou W, Xu J, Li J, Li N, Xu J, You C, Wang C, Tian M. Rosuvastatin Nanomicelles Target Neuroinflammation and Improve Neurological Deficit in a Mouse Model of Intracerebral Hemorrhage. Int J Nanomedicine 2021; 16:2933-2947. [PMID: 33907400 PMCID: PMC8068519 DOI: 10.2147/ijn.s294916] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 04/01/2021] [Indexed: 02/05/2023] Open
Abstract
Background Intracerebral hemorrhage (ICH), a devastating subtype of stroke, has a poor prognosis. However, there is no effective therapy currently available due to its complex pathological progression, in which neuroinflammation plays a pivotal role in secondary brain injury. In this work, the use of statin-loaded nanomicelles to target the neuroinflammation and improve the efficacy was studied in a mouse model of ICH. Methods Rosuvastatin-loaded nanomicelles were prepared by a co-solvent evaporation method using polyethylene glycol-poly(ε-caprolactone) (PEG-PCL) copolymer as a carrier. The prepared nanomicelles were characterized by transmission electron microscopy (TEM) and dynamic light scattering (DLS), and then in vitro and in vivo studies were performed. Results TEM shows that the nanomicelles are spherical with a diameter of about 19.41 nm, and DLS shows that the size, zeta potential, and polymer dispersity index of the nanomicelles were 23.37 nm, −19.2 mV, and 0.221, respectively. The drug loading content is 8.28%. The in vivo study showed that the nanomicelles significantly reduced neuron degeneration, inhibited the inflammatory cell infiltration, reduced the brain edema, and improved neurological deficit. Furthermore, it was observed that the nanomicelles promoted the polarization of microglia/macrophages to M2 phenotype, and also the expression of the proinflammatory cytokines, such as IL-1β and TNF-α, was significantly down-regulated, while the expression of the anti-inflammatory cytokine IL-10 was significantly up-regulated. The related mechanism was proposed and discussed. Conclusion The nanomicelles treatment suppressed the neuroinflammation that might contribute to the promoted nerve functional recovery of the ICH mouse, making it potential to be applied in clinic.
Collapse
Affiliation(s)
- Liu Zi
- Neurosurgery Research Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Wencheng Zhou
- Neurosurgery Research Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jiake Xu
- Neurosurgery Research Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Junshu Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Ning Li
- Department of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jianguo Xu
- Neurosurgery Research Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,West China Brain Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Chao You
- Neurosurgery Research Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,West China Brain Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Chengwei Wang
- Neurosurgery Research Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Meng Tian
- Neurosurgery Research Laboratory, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,West China Brain Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
34
|
Li Y, Zhang X, Liu X, Pan W, Li N, Tang B. Intelligent stimuli-responsive nano immunomodulators for cancer immunotherapy. Chem Sci 2021; 12:3130-3145. [PMID: 34164080 PMCID: PMC8179382 DOI: 10.1039/d0sc06557a] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/01/2021] [Indexed: 12/17/2022] Open
Abstract
Cancer immunotherapy is a revolutionary treatment method in oncology, which uses a human's own immune system against cancer. Many immunomodulators that trigger an immune response have been developed and applied in cancer immunotherapy. However, there is the risk of causing an excessive immune response upon directly injecting common immunomodulators into the human body to trigger an immune response. Therefore, the development of intelligent stimuli-responsive immunomodulators to elicit controlled immune responses in cancer immunotherapy is of great significance. Nanotechnology offers the possibility of designing smart nanomedicine to amplify the antitumor response in a safe and effective manner. Progress relating to intelligent stimuli-responsive nano immunomodulators for cancer immunotherapy is highlighted as a new creative direction in the field. Considering the clinical demand for cancer immunotherapy, we put forward some suggestions for constructing new intelligent stimuli-responsive nano immunomodulators, which will advance the development of cancer immunotherapy.
Collapse
Affiliation(s)
- Yanhua Li
- College of Chemistry, Chemical Engineering and Materials Science Key Laboratory of Molecular and Nano Probes, Ministry of Education Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Xia Zhang
- College of Chemistry, Chemical Engineering and Materials Science Key Laboratory of Molecular and Nano Probes, Ministry of Education Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Xiaohan Liu
- College of Chemistry, Chemical Engineering and Materials Science Key Laboratory of Molecular and Nano Probes, Ministry of Education Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science Key Laboratory of Molecular and Nano Probes, Ministry of Education Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science Key Laboratory of Molecular and Nano Probes, Ministry of Education Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science Key Laboratory of Molecular and Nano Probes, Ministry of Education Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| |
Collapse
|
35
|
Jiang L, Jung S, Zhao J, Kasinath V, Ichimura T, Joseph J, Fiorina P, Liss AS, Shah K, Annabi N, Joshi N, Akama TO, Bromberg JS, Kobayashi M, Uchimura K, Abdi R. Simultaneous targeting of primary tumor, draining lymph node, and distant metastases through high endothelial venule-targeted delivery. NANO TODAY 2021; 36:101045. [PMID: 33391389 PMCID: PMC7774643 DOI: 10.1016/j.nantod.2020.101045] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Cancer patients with malignant involvement of tumor-draining lymph nodes (TDLNs) and distant metastases have the poorest prognosis. A drug delivery platform that targets the primary tumor, TDLNs, and metastatic niches simultaneously, remains to be developed. Here, we generated a novel monoclonal antibody (MHA112) against peripheral node addressin (PNAd), a family of glycoproteins expressed on high endothelial venules (HEVs), which are present constitutively in the lymph nodes (LNs) and formed ectopically in the tumor stroma. MHA112 was endocytosed by PNAd-expressing cells, where it passed through the lysosomes. MHA112 conjugated antineoplastic drug Paclitaxel (Taxol) (MHA112-Taxol) delivered Taxol effectively to the HEV-containing tumors, TDLNs, and metastatic lesions. MHA112-Taxol treatment significantly reduced primary tumor size as well as metastatic lesions in a number of mouse and human tumor xenografts tested. These data, for the first time, indicate that human metastatic lesions contain HEVs and provide a platform that permits simultaneous targeted delivery of antineoplastic drugs to the three key sites of primary tumor, TDLNs, and metastases.
Collapse
Affiliation(s)
- Liwei Jiang
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sungwook Jung
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jing Zhao
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Vivek Kasinath
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Takaharu Ichimura
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - John Joseph
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Paolo Fiorina
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew S. Liss
- Department of Surgery and the Andrew L. Warshaw, MD Institute for Pancreatic Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Khalid Shah
- Center for Stem Cell Therapeutics and Imaging, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard medical School, Boston, MA, 02115, USA
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Nitin Joshi
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tomoya O. Akama
- Department of Pharmacology, Kansai Medical University, Osaka, 570-8506, Japan
| | - Jonathan S. Bromberg
- Departments of Surgery and Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Motohiro Kobayashi
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Kenji Uchimura
- Department of Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
- CNRS, UMR 8576, Unit of Glycobiology Structures and Functions, University of Lille, F-59000 Lille, France
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
36
|
Environmental control of mammary carcinoma cell expansion by acidification and spheroid formation in vitro. Sci Rep 2020; 10:21959. [PMID: 33319820 PMCID: PMC7738540 DOI: 10.1038/s41598-020-78989-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 12/01/2020] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is the leading cause of cancer death among women worldwide. Like other cancers, mammary carcinoma progression involves acidification of the tumor microenvironment, which is an important factor for cancer detection and treatment strategies. However, the effects of acidity on mammary carcinoma cell morphology and phenotype have not been thoroughly characterized. Here, we evaluated fundamental effects of environmental acidification on mammary carcinoma cells in standard two-dimensional cultures and three-dimensional spheroids. Acidification decreased overall mammary carcinoma cell viability, while increasing their resistance to the anthracycline doxorubicin. Environmental acidification also increased extracellular vesicle production by mammary carcinoma cells. Conditioned media containing these vesicles appeared to increase fibroblast motility. Acidification also increased mammary carcinoma cell motility when cultured with fibroblasts in spheroids. Taken together, results from this study suggest that environmental acidification induces drug resistance and extracellular vesicle production by mammary carcinoma cells that promote tumor expansion.
Collapse
|
37
|
Li J, Burgess DJ. Nanomedicine-based drug delivery towards tumor biological and immunological microenvironment. Acta Pharm Sin B 2020; 10:2110-2124. [PMID: 33304781 PMCID: PMC7714990 DOI: 10.1016/j.apsb.2020.05.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/15/2020] [Accepted: 05/21/2020] [Indexed: 12/17/2022] Open
Abstract
The complex tumor microenvironment is a most important factor in cancer development. The biological microenvironment is composed of a variety of barriers including the extracellular matrix and associated cells such as endothelia cells, pericytes, and cancer-associated fibroblasts. Different strategies can be utilized to enhance nanoparticle-based drug delivery and distribution into tumor tissues addressing the extracellular matrix or cellular components. In addition to the biological microenvironment, the immunological conditions around the tumor tissue can be very complicated and cancer cells have various ways of evading immune surveillance. Nanoparticle drug delivery systems can enhance cancer immunotherapy by tuning the immunological response and memory of various immune cells such as T cells, B cells, macrophages, and dendritic cells. In this review, the main components in the tumor biological and immunological environment are discussed. The focus is on recent advances in nanoparticle-based drug delivery systems towards targets within the tumor microenvironment to improve cancer chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Jin Li
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269-3092, USA
| | - Diane J. Burgess
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269-3092, USA
| |
Collapse
|
38
|
Ahmadabad LE, Kalantari FS, Liu H, Hasan A, Gamasaee NA, Edis Z, Attar F, Ale-Ebrahim M, Rouhollah F, Babadaei MMN, Sharifi M, Shahpasand K, Akhtari K, Falahati M, Cai Y. Hydrothermal method-based synthesized tin oxide nanoparticles: Albumin binding and antiproliferative activity against K562 cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111649. [PMID: 33321685 DOI: 10.1016/j.msec.2020.111649] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/18/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022]
Abstract
The interaction of nanoparticles with protein and cells may provide important information regarding their biomedical implementations. Herein, after synthesis of tin oxide (SnO2) nanoparticles by hydrothermal method, their interaction with human serum albumin (HSA) was evaluated by multispectroscopic and molecular docking (MD) approaches. Furthermore, the selective antiproliferative impact of SnO2 nanoparticles against leukemia K562 cells was assessed by different cellular assays, whereas lymphocytes were used as control cells. TEM, DLS, zeta potential and XRD techniques showed that crystalline SnO2 nanoparticles have a size of less than 50 nm with a good colloidal stability. Fluorescence and CD spectroscopy analysis indicated that the HSA undergoes some slight conformational changes after interaction with SnO2 nanoparticles, whereas the secondary structure of HSA remains intact. Moreover, MD outcomes revealed that the charged residues of HSA preferentially bind to SnO2 nanoclusters in the binding pocket. Antiproliferative examinations displayed that SnO2 nanoparticles can selectively cause the mortality of K562 cells through induction of cell membrane leakage, activation of caspase-9, -8, -3, down regulation of Bcl-2 mRNA, the elevation of ROS level, S phase arrest, and apoptosis. In conclusion, this data may indicate that SnO2 nanoparticles can be used as promising particles to be integrated into therapeutic platforms.
Collapse
Affiliation(s)
- Leila Ebrahimi Ahmadabad
- Department of Cellular and Molecular Biology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Firoozeh Samia Kalantari
- Department of Cellular and Molecular Biology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hui Liu
- College of Pharmacy, Jinan University, Guangzhou, Guangdong 510632, China
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Centre, Qatar University, Doha 2713, Qatar.
| | - Niusha Abbasi Gamasaee
- Department of Genetics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zehra Edis
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Ajman University, PO Box 346, Ajman, United Arab Emirates
| | - Farnoosh Attar
- Department of Food Toxicology, Research Center of Food Technology and Agricultural Products, Standard Research Institute (SRI), Karaj, Iran
| | - Mahsa Ale-Ebrahim
- Department of Physiology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Rouhollah
- Department of Cellular and Molecular Biology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Mahdi Nejadi Babadaei
- Department of Molecular Genetics, Faculty of Biological Science, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Majid Sharifi
- Department of Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Koorosh Shahpasand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology (RI-SCBT), Tehran, Iran
| | - Keivan Akhtari
- Department of Physics, University of Kurdistan, Sanandaj, Iran
| | - Mojtaba Falahati
- Department of Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Yu Cai
- College of Pharmacy, Jinan University, Guangzhou, Guangdong 510632, China; Cancer Institute of Jinan University, Guangzhou, Guangdong 510632, China; International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), School of Pharmacy, Jinan University, Guangzhou, Guangdong 510632, China.
| |
Collapse
|
39
|
Karaosmanoglu S, Zhou M, Shi B, Zhang X, Williams GR, Chen X. Carrier-free nanodrugs for safe and effective cancer treatment. J Control Release 2020; 329:805-832. [PMID: 33045313 DOI: 10.1016/j.jconrel.2020.10.014] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/19/2022]
Abstract
Clinical applications of many anti-cancer drugs are restricted due to their hydrophobic nature, requiring use of harmful organic solvents for administration, and poor selectivity and pharmacokinetics resulting in off-target toxicity and inefficient therapies. A wide variety of carrier-based nanoparticles have been developed to tackle these issues, but such strategies often fail to encapsulate drug efficiently and require significant amounts of inorganic and/or organic nanocarriers which may cause toxicity problems in the long term. Preparation of nano-formulations for the delivery of water insoluble drugs without using carriers is thus desired, requiring elegantly designed strategies for products with high quality, stability and performance. These strategies include simple self-assembly or involving chemical modifications via coupling drugs together or conjugating them with various functional molecules such as lipids, carbohydrates and photosensitizers. During nanodrugs synthesis, insertion of redox-responsive linkers and tumor targeting ligands endows them with additional characteristics like on-target delivery, and conjugation with immunotherapeutic reagents enhances immune response alongside therapeutic efficacy. This review aims to summarize the methods of making carrier-free nanodrugs from hydrophobic drug molecules, evaluating their performance, and discussing the advantages, challenges, and future development of these strategies.
Collapse
Affiliation(s)
- Sena Karaosmanoglu
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, King's Buildings, Mayfield Road, Edinburgh EH9 3JL, UK
| | - Mengjiao Zhou
- Department of Pharmacology, School of Pharmacy, Nantong University, 226000, Nantong, Jiangsu, PR China
| | - Bingyang Shi
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Xiujuan Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, Jiangsu, PR China.
| | - Gareth R Williams
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| | - Xianfeng Chen
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, King's Buildings, Mayfield Road, Edinburgh EH9 3JL, UK.
| |
Collapse
|
40
|
Li Y, He X, Fan L, Zhang X, Xu Y, Xu X. Identification of a novel immune prognostic model in gastric cancer. Clin Transl Oncol 2020; 23:846-855. [PMID: 32857339 DOI: 10.1007/s12094-020-02478-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/10/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE The tumor immune microenvironment (TIME) is now considered as an important factor during gastric cancer (GC) development. This study identified a novel immune-related risk model for predicting prognosis and assessing the immune status of GC patients. METHODS Transcriptomic data were obtained from the TCGA database. The differential expressed immune-related genes (IRGs) were identified through the ImmPort portal. Enrichment analysis was performed to explore the potential molecular mechanism of these IRGs. By the Cox regression analysis, we constructed the immune prognostic model. Then, the association between the model and the immune microenvironment was estimated. The model was validated in the GSE84433 dataset. RESULTS Totally, we identified 222 differentially expressed IRGs. These IRGs were closely correlated with immune response and immune signaling pathways. Through the Cox regression analysis, we developed the immune prognostic model based on the expression of seven IRGs (CXCL3, NOX4, PROC, FAM19A4, RNASE2, IGHD2-15, CGB5). Patients were stratified into two groups according to immune-related risk scores. Survival analysis indicated that the prognosis of high-risk patients was poorer than low-risk patients. Moreover, the immune-related risk score was an independent prognostic biomarker. More importantly, we found that the infiltration level of immunosuppressive cells and the expression of inhibitory immune checkpoints were higher in high-risk patients. The immune microenvironment tended to be a suppressive status in patients with high-risk scores. CONCLUSION This study demonstrated that our model had predictive value for prognosis and the TIME in GC. It might be a robust tool to improve personalized patient management.
Collapse
Affiliation(s)
- Y Li
- Cancer Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - X He
- Cancer Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - L Fan
- Cancer Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - X Zhang
- Cancer Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Y Xu
- Cancer Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - X Xu
- Cancer Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
| |
Collapse
|
41
|
Liu G, Gu A, E M. [Mechanism and Research Progress of Microbiome in the Development of Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:948-953. [PMID: 32819055 PMCID: PMC7679216 DOI: 10.3779/j.issn.1009-3419.2020.101.38] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
肺癌是我国最常见的、预后较差的恶性肿瘤之一,大多数患者确诊时即为晚期。有研究表明肺癌患者的微生态特征有别于健康人群,呼吸道的微生物可以通过多种机制影响肺癌的发生发展。近年来微生物组学与疾病相关性研究已成为继人类基因组计划又一研究热点,然而目前有关肺癌与呼吸道微生物组特征的研究相对较少,因此,需要更深入地探讨肺癌与微生物菌群间的潜在联系,通过研究呼吸道微生物在肺癌发生发展中的作用机制,以期在为肺癌的临床诊治、预后评估提供更明确的科学依据。本文对目前关于微生物菌群与肺癌相关研究进行综述,并为临床上诊断与治疗肺癌提供新的思路。
Collapse
Affiliation(s)
- Guohui Liu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin 150040, China
| | - Anxin Gu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin 150040, China
| | - Mingyan E
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin 150040, China
| |
Collapse
|
42
|
Liu Y, Guo J, Huang L. Modulation of tumor microenvironment for immunotherapy: focus on nanomaterial-based strategies. Am J Cancer Res 2020; 10:3099-3117. [PMID: 32194857 PMCID: PMC7053194 DOI: 10.7150/thno.42998] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 01/19/2020] [Indexed: 02/07/2023] Open
Abstract
Recent advances in the field of immunotherapy have profoundly opened up the potential for improved cancer therapy and reduced side effects. However, the tumor microenvironment (TME) is highly immunosuppressive, therefore, clinical outcomes of currently available cancer immunotherapy are still poor. Recently, nanomaterial-based strategies have been developed to modulate the TME for robust immunotherapeutic responses. In this review, the immunoregulatory cell types (cells relating to the regulation of immune responses) inside the TME in terms of stimulatory and suppressive roles are described, and the technologies used to identify and quantify these cells are provided. In addition, recent examples of nanomaterial-based cancer immunotherapy are discussed, with particular emphasis on those designed to overcome barriers caused by the complexity and diversity of TME.
Collapse
|
43
|
Qin Y, Liu T, Guo M, Liu Y, Liu C, Chen Y, Qu D. Mild-heat-inducible sequentially released liposomal complex remodels the tumor microenvironment and reinforces anti-breast-cancer therapy. Biomater Sci 2020; 8:3916-3925. [PMID: 32555847 DOI: 10.1039/d0bm00498g] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Heat-responsive drug release helps celastrol & STS-coloaded liposome activate the cascade of TME normalization and enhances the anti-tumor efficacy.
Collapse
Affiliation(s)
- Yue Qin
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
- Nanjing University of Chinese Medicine
- Nanjing 210028
- China
- Jiangsu Provincial Academy of Traditional Chinese Medicine
| | - Tingting Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
- Nanjing University of Chinese Medicine
- Nanjing 210028
- China
| | - Mengfei Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
- Nanjing University of Chinese Medicine
- Nanjing 210028
- China
| | - Yuping Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
- Nanjing University of Chinese Medicine
- Nanjing 210028
- China
- Jiangsu Provincial Academy of Traditional Chinese Medicine
| | - Congyan Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
- Nanjing University of Chinese Medicine
- Nanjing 210028
- China
- Jiangsu Provincial Academy of Traditional Chinese Medicine
| | - Yan Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
- Nanjing University of Chinese Medicine
- Nanjing 210028
- China
- Jiangsu Provincial Academy of Traditional Chinese Medicine
| | - Ding Qu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine
- Nanjing University of Chinese Medicine
- Nanjing 210028
- China
- Jiangsu Provincial Academy of Traditional Chinese Medicine
| |
Collapse
|